Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Proc Natl Acad Sci U S A ; 114(7): 1649-1654, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28143939

ABSTRACT

The secreted Wnt signaling molecules are essential to the coordination of cell-fate decision making in multicellular organisms. In adult animals, the secreted Wnt proteins are critical for tissue regeneration and frequently contribute to cancer. Small molecules that disable the Wnt acyltransferase Porcupine (Porcn) are candidate anticancer agents in clinical testing. Here we have systematically assessed the effects of the Porcn inhibitor (WNT-974) on the regeneration of several tissue types to identify potentially unwanted chemical effects that could limit the therapeutic utility of such agents. An unanticipated observation from these studies is proregenerative responses in heart muscle induced by systemic chemical suppression of Wnt signaling. Using in vitro cultures of several cell types found in the heart, we delineate the Wnt signaling apparatus supporting an antiregenerative transcriptional program that includes a subunit of the nonfibrillar collagen VI. Similar to observations seen in animals exposed to WNT-974, deletion of the collagen VI subunit, COL6A1, has been shown to decrease aberrant remodeling and fibrosis in infarcted heart tissue. We demonstrate that WNT-974 can improve the recovery of heart function after left anterior descending coronary artery ligation by mitigating adverse remodeling of infarcted tissue. Injured heart tissue exposed to WNT-974 exhibits decreased scarring and reduced Col6 production. Our findings support the development of Porcn inhibitors as antifibrotic agents that could be exploited to promote heart repair following injury.


Subject(s)
Acyltransferases/antagonists & inhibitors , Atrial Remodeling/drug effects , Enzyme Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Myocardial Infarction/prevention & control , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Atrial Remodeling/genetics , Cells, Cultured , Collagen Type VI/genetics , Collagen Type VI/metabolism , Enzyme Inhibitors/chemistry , Gene Expression/drug effects , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Molecular Structure , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Pyrazines/chemistry , Pyrazines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Regeneration/drug effects , Regeneration/genetics , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
2.
Nat Chem Biol ; 12(4): 218-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26829472

ABSTRACT

A hallmark of targeted cancer therapies is selective toxicity among cancer cell lines. We evaluated results from a viability screen of over 200,000 small molecules to identify two chemical series, oxalamides and benzothiazoles, that were selectively toxic at low nanomolar concentrations to the same 4 of 12 human lung cancer cell lines. Sensitive cell lines expressed cytochrome P450 (CYP) 4F11, which metabolized the compounds into irreversible inhibitors of stearoyl CoA desaturase (SCD). SCD is recognized as a promising biological target in cancer and metabolic disease. However, SCD is essential to sebocytes, and accordingly SCD inhibitors cause skin toxicity. Mouse sebocytes did not activate the benzothiazoles or oxalamides into SCD inhibitors, providing a therapeutic window for inhibiting SCD in vivo. We thus offer a strategy to target SCD in cancer by taking advantage of high CYP expression in a subset of tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Drug Discovery/methods , Lung Neoplasms/enzymology , Oxamic Acid/analogs & derivatives , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Benzothiazoles/pharmacokinetics , Benzothiazoles/therapeutic use , Benzothiazoles/toxicity , Cell Line, Tumor , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 4 , Female , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, SCID , Molecular Structure , Molecular Targeted Therapy , Oxamic Acid/pharmacokinetics , Oxamic Acid/pharmacology , Oxamic Acid/therapeutic use , Oxamic Acid/toxicity , Protein Binding , Sebaceous Glands/drug effects , Sebaceous Glands/enzymology , Sebaceous Glands/pathology , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem Lett ; 26(24): 5891-5895, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27876319

ABSTRACT

The acyltransferase Porcupine (Porcn) is essential for the secretion of Wnt proteins which contribute to embryonic development, tissue regeneration, and tumorigenesis. We have previously discovered four molecular scaffolds harboring Porcn-inhibitory activity. Comparison of their structures led to the identification of a general scaffold that can be readily assembled by modular synthesis. We report herein the development of a triazole version of this new class of Porcn inhibitors. This study yielded IWP-O1, a Porcn inhibitor with an EC50 value of 80pM in a cultured cell reporter assay of Wnt signaling. Additionally, IWP-O1 has significantly improved metabolic stability over our previously reported Porcn inhibitors.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Triazoles/pharmacology , Acyltransferases , Dose-Response Relationship, Drug , Humans , Membrane Proteins/metabolism , Molecular Structure , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
4.
Proc Natl Acad Sci U S A ; 110(24): 9728-33, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23716694

ABSTRACT

The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are elevated in maple syrup urine disease, heart failure, obesity, and type 2 diabetes. BCAA homeostasis is controlled by the mitochondrial branched-chain α-ketoacid dehydrogenase complex (BCKDC), which is negatively regulated by the specific BCKD kinase (BDK). Here, we used structure-based design to develop a BDK inhibitor, (S)-α-chloro-phenylpropionic acid [(S)-CPP]. Crystal structures of the BDK-(S)-CPP complex show that (S)-CPP binds to a unique allosteric site in the N-terminal domain, triggering helix movements in BDK. These conformational changes are communicated to the lipoyl-binding pocket, which nullifies BDK activity by blocking its binding to the BCKDC core. Administration of (S)-CPP to mice leads to the full activation and dephosphorylation of BCKDC with significant reduction in plasma BCAA concentrations. The results buttress the concept of targeting mitochondrial BDK as a pharmacological approach to mitigate BCAA accumulation in metabolic diseases and heart failure.


Subject(s)
Mitochondrial Proteins/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinases/chemistry , Protein Structure, Tertiary , Allosteric Regulation , Animals , Binding Sites/genetics , Chromatography, Liquid , Crystallography, X-Ray , Isoleucine/blood , Isoleucine/metabolism , Kinetics , Leucine/blood , Leucine/metabolism , Male , Mice , Mice, Inbred ICR , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Models, Molecular , Molecular Structure , Mutation , Phenylpropionates/chemistry , Phenylpropionates/metabolism , Phenylpropionates/pharmacology , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Tandem Mass Spectrometry , Valine/blood , Valine/metabolism
5.
J Biol Chem ; 289(7): 4432-43, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24356970

ABSTRACT

Pyruvate dehydrogenase kinase isoforms (PDKs 1-4) negatively regulate activity of the mitochondrial pyruvate dehydrogenase complex by reversible phosphorylation. PDK isoforms are up-regulated in obesity, diabetes, heart failure, and cancer and are potential therapeutic targets for these important human diseases. Here, we employed a structure-guided design to convert a known Hsp90 inhibitor to a series of highly specific PDK inhibitors, based on structural conservation in the ATP-binding pocket. The key step involved the substitution of a carbonyl group in the parent compound with a sulfonyl in the PDK inhibitors. The final compound of this series, 2-[(2,4-dihydroxyphenyl)sulfonyl]isoindoline-4,6-diol, designated PS10, inhibits all four PDK isoforms with IC50 = 0.8 µM for PDK2. The administration of PS10 (70 mg/kg) to diet-induced obese mice significantly augments pyruvate dehydrogenase complex activity with reduced phosphorylation in different tissues. Prolonged PS10 treatments result in improved glucose tolerance and notably lessened hepatic steatosis in the mouse model. The results support the pharmacological approach of targeting PDK to control both glucose and fat levels in obesity and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Enzyme Inhibitors , Fatty Liver/drug therapy , Isoindoles/chemistry , Isoindoles/pharmacology , Obesity/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sulfones/chemistry , Sulfones/pharmacology , Animals , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Drug Delivery Systems , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fatty Liver/enzymology , Fatty Liver/genetics , Fatty Liver/pathology , HSP90 Heat-Shock Proteins , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Obese , Obesity/enzymology , Obesity/genetics , Obesity/pathology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
6.
J Biol Chem ; 289(30): 20583-93, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24895126

ABSTRACT

The mitochondrial branched-chain α-ketoacid dehydrogenase complex (BCKDC) is negatively regulated by reversible phosphorylation.BCKDC kinase (BDK) inhibitors that augment BCKDC flux have been shown to reduce branched-chain amino acid (BCAA) concentrations in vivo. In the present study, we employed high-throughput screens to identify compound 3,6- dichlorobenzo[b]thiophene-2-carboxylic acid (BT2) as a novel BDK inhibitor (IC(50) = 3.19 µM). BT2 binds to the same site in BDK as other known allosteric BDK inhibitors, including (S)-α-cholorophenylproprionate ((S)-CPP). BT2 binding to BDK triggers helix movements in the N-terminal domain, resulting in the dissociation of BDK from the BCKDC accompanied by accelerated degradation of the released kinase in vivo. BT2 shows excellent pharmacokinetics (terminal T(1/2) = 730 min) and metabolic stability (no degradation in 240 min), which are significantly better than those of (S)-CPP. BT2, its analog 3-chloro-6-fluorobenzo[ b]thiophene-2-carboxylic acid (BT2F), and a prodrug of BT2 (i.e. N-(4-acetamido-1,2,5-oxadiazol-3-yl)-3,6-dichlorobenzo[ b]thiophene-2-carboxamide (BT3)) significantly increase residual BCKDC activity in cultured cells and primary hepatocytes from patients and a mouse model of maple syrup urine disease. Administration of BT2 at 20 mg/kg/day to wild-type mice for 1 week leads to nearly complete dephosphorylation and maximal activation of BCKDC in heart, muscle, kidneys, and liver with reduction in plasma BCAA concentrations. The availability of benzothiophene carboxylate derivatives as stable BDK inhibitors may prove useful for the treatment of metabolic disease caused by elevated BCAA concentrations.


Subject(s)
Enzyme Inhibitors/pharmacology , Hepatocytes/enzymology , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Ketoglutarate Dehydrogenase Complex/metabolism , Proteolysis/drug effects , Thiophenes/pharmacology , Allosteric Regulation/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Stability/drug effects , Enzyme Stability/genetics , Hepatocytes/pathology , Humans , Ketoglutarate Dehydrogenase Complex/genetics , Mice , Mice, Knockout , Thiophenes/pharmacokinetics
7.
Proc Natl Acad Sci U S A ; 109(42): 17016-21, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027932

ABSTRACT

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the hippocampal dentate gyrus. We have further found that chemicals having efficacy in this in vivo screening assay also protect dopaminergic neurons of the substantia nigra following exposure to the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a mouse model of Parkinson disease. Here, we provide evidence that an active analog of P7C3, known as P7C3A20, protects ventral horn spinal cord motor neurons from cell death in the G93A-SOD1 mutant mouse model of amyotrophic lateral sclerosis (ALS). P7C3A20 is efficacious in this model when administered at disease onset, and protection from cell death correlates with preservation of motor function in assays of walking gait and in the accelerating rotarod test. The prototypical member of this series, P7C3, delays disease progression in G93A-SOD1 mice when administration is initiated substantially earlier than the expected time of symptom onset. Dimebon, an antihistaminergic drug with significantly weaker proneurogenic and neuroprotective efficacy than P7C3, confers no protection in this ALS model. We propose that the chemical scaffold represented by P7C3 and P7C3A20 may provide a basis for the discovery and optimization of pharmacologic agents for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/prevention & control , Carbazoles/pharmacology , Motor Neurons/cytology , Neuroprotective Agents/pharmacology , Spinal Cord/cytology , Animals , Carbazoles/chemical synthesis , Carbazoles/chemistry , Carbazoles/pharmacokinetics , Indoles/pharmacokinetics , Indoles/pharmacology , Mice , Motor Activity/drug effects , Motor Activity/physiology , Motor Neurons/drug effects , Polymerase Chain Reaction , Rotarod Performance Test , Spinal Cord/drug effects
8.
Proc Natl Acad Sci U S A ; 109(42): 17010-5, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027934

ABSTRACT

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the dentate gyrus. Here, we provide evidence that P7C3 also protects mature neurons in brain regions outside of the hippocampus. P7C3 blocks 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated cell death of dopaminergic neurons in the substantia nigra of adult mice, a model of Parkinson disease (PD). Dose-response studies show that the P7C3 analog P7C3A20 blocks cell death with even greater potency and efficacy, which parallels the relative potency and efficacy of these agents in blocking apoptosis of newborn neural precursor cells of the dentate gyrus. P7C3 and P7C3A20 display similar relative effects in blocking 1-methyl-4-phenylpyridinium (MPP(+))-mediated death of dopaminergic neurons in Caenorhabditis elegans, as well as in preserving C. elegans mobility following MPP(+) exposure. Dimebon, an antihistaminergic drug that is weakly proneurogenic and neuroprotective in the dentate gyrus, confers no protection in either the mouse or the worm models of PD. We further demonstrate that the hippocampal proneurogenic efficacy of eight additional analogs of P7C3 correlates with their protective efficacy in MPTP-mediated neurotoxicity. In vivo screening of P7C3 analogs for proneurogenic efficacy in the hippocampus may thus provide a reliable means of predicting neuroprotective efficacy. We propose that the chemical scaffold represented by P7C3 and P7C3A20 provides a basis for optimizing and advancing pharmacologic agents for the treatment of patients with PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/antagonists & inhibitors , Carbazoles/pharmacology , Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Substantia Nigra/cytology , Animals , Apoptosis/drug effects , Caenorhabditis elegans , Carbazoles/chemical synthesis , Carbazoles/chemistry , Carbazoles/pharmacokinetics , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/drug effects , Indoles/pharmacokinetics , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Molecular Structure , Substantia Nigra/drug effects
9.
J Med Chem ; 67(11): 9277-9301, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38804887

ABSTRACT

We recently discovered a novel N-aryl tetracyclic dicarboximide MM0299 (1) with robust activity against glioma stem-like cells that potently and selectively inhibits lanosterol synthase leading to the accumulation of the toxic shunt metabolite 24(S),25-epoxycholesterol. Herein, we delineate a systematic and comprehensive SAR study that explores the structural space surrounding the N-aryl tetracyclic dicarboximide scaffold. A series of 100 analogs were synthesized and evaluated for activity against the murine glioma stem-like cell line Mut6 and for metabolic stability in mouse liver S9 fractions. This study led to several analogs with single-digit nanomolar activity in Mut6 glioblastoma cells that were metabolically stable in S9 fractions. In vivo pharmacokinetic analysis of selected analogs identified compound 52a (IC50 = 63 nM; S9 T1/2 > 240 min) which was orally available (39% plasma; 58% brain) and displayed excellent brain exposure. Chronic oral dosing of 52a during a 2-week tolerability study indicated no adverse effect on body weight nor signs of hematologic, liver, or kidney toxicity.


Subject(s)
Glioma , Neoplastic Stem Cells , Animals , Mice , Structure-Activity Relationship , Glioma/drug therapy , Glioma/pathology , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Drug Discovery , Male , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology
10.
J Am Chem Soc ; 133(5): 1428-37, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21210688

ABSTRACT

Degeneration of the hippocampus is associated with Alzheimer's disease and occurs very early in the progression of the disease. Current options for treating the cognitive symptoms associated with Alzheimer's are inadequate, giving urgency to the search for novel therapeutic strategies. Pharmacologic agents that safely enhance hippocampal neurogenesis may provide new therapeutic approaches. We discovered the first synthetic molecule, named P7C3, which protects newborn neurons from apoptotic cell death, and thus promotes neurogenesis in mice and rats in the subgranular zone of the hippocampal dentate gyrus, the site of normal neurogenesis in adult mammals. We describe the results of a medicinal chemistry campaign to optimize the potency, toxicity profile, and stability of P7C3. Systematic variation of nearly every position of the lead compound revealed elements conducive toward increases in activity and regions subject to modification. We have discovered compounds that are orally available, nontoxic, stable in mice, rats, and cell culture, and capable of penetrating the blood-brain barrier. The most potent compounds are active at nanomolar concentrations. Finally, we have identified derivatives that may facilitate mode-of-action studies through affinity chromatography or photo-cross-linking.


Subject(s)
Carbazoles/chemistry , Carbazoles/pharmacology , Drug Discovery/methods , Neurogenesis/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Animals , Carbazoles/therapeutic use , Carbazoles/toxicity , Dose-Response Relationship, Drug , Drug Stability , HeLa Cells , Humans , Male , Mice , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/toxicity , Structure-Activity Relationship
11.
J Med Chem ; 62(10): 5217-5241, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31070915

ABSTRACT

Despite advances in targeted anticancer therapies, there are still no small-molecule-based therapies available that specifically target colorectal cancer (CRC) development and progression, the second leading cause of cancer deaths. We previously disclosed the discovery of truncating adenomatous polyposis coli (APC)-selective inhibitor 1 (TASIN-1), a small molecule that specifically targets colorectal cancer cells lines with truncating mutations in the adenomatous polyposis coli (APC) tumor suppressor gene through inhibition of cholesterol biosynthesis. Here, we report a medicinal chemistry evaluation of a collection of TASIN analogues and activity against colon cancer cell lines and an isogenic cell line pair reporting on the status of APC-dependent selectivity. A number of potent and selective analogues were identified, including compounds with good metabolic stability and pharmacokinetic properties. The compounds reported herein represent a first-in-class genotype-selective series that specifically target apc mutations present in the majority of CRC patients and serve as a translational platform toward a targeted therapy for colon cancer.


Subject(s)
Adenomatous Polyposis Coli Protein/drug effects , Adenomatous Polyposis Coli/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Piperidines/antagonists & inhibitors , Sulfonamides/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/genetics , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Drug Design , Drug Discovery , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred ICR , Mutation/drug effects , Protein Binding , Structure-Activity Relationship
12.
EBioMedicine ; 39: 145-158, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30528456

ABSTRACT

BACKGROUND: Chromosomal translocation-induced expression of the chromatin modifying oncofusion protein MLL-AF9 promotes acute myelocytic leukemia (AML). Whereas WNT/ß-catenin signaling has previously been shown to support MLL-AF9-driven leukemogenesis, the mechanism underlying this relationship remains unclear. METHODS: We used two novel small molecules targeting WNT signaling as well as a genetically modified mouse model that allow targeted deletion of the WNT protein chaperone Wntless (WLS) to evaluate the role of WNT signaling in AML progression. ATAC-seq and transcriptome profiling were deployed to understand the cellular consequences of disrupting a WNT signaling in leukemic initiating cells (LICs). FINDINGS: We identified Six1 to be a WNT-controlled target gene in MLL-AF9-transformed leukemic initiating cells (LICs). MLL-AF9 alters the accessibility of Six1 DNA to the transcriptional effector TCF7L2, a transducer of WNT/ß-catenin gene expression changes. Disruption of WNT/SIX1 signaling using inhibitors of the Wnt signaling delays the development of AML. INTERPRETATION: By rendering TCF/LEF-binding elements controlling Six1 accessible to TCF7L2, MLL-AF9 promotes WNT/ß-catenin-dependent growth of LICs. Small molecules disrupting WNT/ß-catenin signaling block Six1 expression thereby disrupting leukemia driven by MLL fusion proteins.


Subject(s)
Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway/drug effects , Animals , HEK293 Cells , HL-60 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/genetics , THP-1 Cells , Transcription Factor 7-Like 2 Protein/metabolism
13.
J Med Chem ; 60(3): 1142-1150, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28085286

ABSTRACT

Pyruvate dehydrogenase kinases 1-4 (PDK1-4) negatively control activity of the pyruvate dehydrogenase complex (PDC) and are up-regulated in obesity, diabetes, heart failure, and cancer. We reported earlier two novel pan-PDK inhibitors PS8 [4-((5-hydroxyisoindolin-2-yl)sulfonyl)benzene-1,3-diol] (1) and PS10 [2-((2,4-dihydroxyphenyl)sulfonyl)isoindoline-4,6-diol] (2) that targeted the ATP-binding pocket in PDKs. Here, we developed a new generation of PDK inhibitors by extending the dihydroxyphenyl sulfonylisoindoline scaffold in 1 and 2 to the entrance region of the ATP-binding pocket in PDK2. The lead inhibitor (S)-3-amino-4-(4-((2-((2,4-dihydroxyphenyl)sulfonyl)isoindolin-5-yl)amino)piperidin-1-yl)-4-oxobutanamide (17) shows a ∼8-fold lower IC50 (58 nM) than 2 (456 nM). In the crystal structure, the asparagine moiety in 17 provides additional interactions with Glu-262 from PDK2. Treatment of diet-induced obese mice with 17 resulted in significant liver-specific augmentation of PDC activity, accompanied by improved glucose tolerance and drastically reduced hepatic steatosis. These findings support 17 as a potential glucose-lowering therapeutic targeting liver for obesity and type 2 diabetes.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Crystallography, X-Ray , Female , Indoles/chemistry , Inhibitory Concentration 50 , Mice , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
14.
mBio ; 8(2)2017 04 04.
Article in English | MEDLINE | ID: mdl-28377530

ABSTRACT

Ebola virus (EBOV) protein VP35 inhibits production of interferon alpha/beta (IFN) by blocking RIG-I-like receptor signaling pathways, thereby promoting virus replication and pathogenesis. A high-throughput screening assay, developed to identify compounds that either inhibit or bypass VP35 IFN-antagonist function, identified five DNA intercalators as reproducible hits from a library of bioactive compounds. Four, including doxorubicin and daunorubicin, are anthracycline antibiotics that inhibit topoisomerase II and are used clinically as chemotherapeutic drugs. These compounds were demonstrated to induce IFN responses in an ATM kinase-dependent manner and to also trigger the DNA-sensing cGAS-STING pathway of IFN induction. These compounds also suppress EBOV replication in vitro and induce IFN in the presence of IFN-antagonist proteins from multiple negative-sense RNA viruses. These findings provide new insights into signaling pathways activated by important chemotherapy drugs and identify a novel therapeutic approach for IFN induction that may be exploited to inhibit RNA virus replication.IMPORTANCE Ebola virus and other emerging RNA viruses are significant but unpredictable public health threats. Therapeutic approaches with broad-spectrum activity could provide an attractive response to such infections. We describe a novel assay that can identify small molecules that overcome Ebola virus-encoded innate immune evasion mechanisms. This assay identified as hits cancer chemotherapeutic drugs, including doxorubicin. Follow-up studies provide new insight into how doxorubicin induces interferon (IFN) responses, revealing activation of both the DNA damage response kinase ATM and the DNA sensor cGAS and its partner signaling protein STING. The studies further demonstrate that the ATM and cGAS-STING pathways of IFN induction are a point of vulnerability not only for Ebola virus but for other RNA viruses as well, because viral innate immune antagonists consistently fail to block these signals. These studies thereby define a novel avenue for therapeutic intervention against emerging RNA viruses.


Subject(s)
Antiviral Agents/pharmacology , DNA Damage/immunology , Ebolavirus/physiology , Immune Evasion/drug effects , Interferons/metabolism , Topoisomerase II Inhibitors/pharmacology , Virus Replication/drug effects , Cell Line , Ebolavirus/immunology , Humans
15.
Cell Metab ; 21(1): 117-25, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25533479

ABSTRACT

Dietary effects on tumor biology can be exploited to unravel cancer vulnerabilities. Here, we present surprising evidence for anti-proliferative action of high-calorie-diet (HCD) feeding on KRAS-driven lung tumors. Tumors of mice that commenced HCD feeding before tumor onset displayed defective unfolded protein response (UPR) and unresolved endoplasmic reticulum (ER) stress. Unresolved ER stress and reduced proliferation are reversed by chemical chaperone treatment. Whole-genome transcriptional analyses revealed FKBP10 as one of the most downregulated chaperones in tumors of the HCD-pre-tumor-onset group. FKBP10 downregulation dampens tumor growth in vitro and in vivo. Providing translational value to these results, we report that FKBP10 is expressed in human KRAS-positive and -negative lung cancers, but not in healthy parenchyma. Collectively, our data shed light on an unexpected anti-tumor action of HCD imposed before tumor onset and identify FKBP10 as a putative therapeutic target to selectively hinder lung cancer.


Subject(s)
Diet , Lung Neoplasms/pathology , Phenylbutyrates/toxicity , ras Proteins/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Down-Regulation , Doxycycline/toxicity , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, SCID , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tacrolimus Binding Proteins/antagonists & inhibitors , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Transplantation, Heterologous , Unfolded Protein Response/drug effects , ras Proteins/genetics
16.
Article in English | MEDLINE | ID: mdl-27158662

ABSTRACT

BACKGROUND: There are currently no therapeutic options for patients with Parkinson's disease that prevent or slow the death of dopaminergic neurons. We have recently identified the novel P7C3 class of neuroprotective molecules that blocks neuron cell death. AIMS: The aim of this study was to determine whether treatment with highly active members of the P7C3 series blocks dopaminergic neuron cell death and associated behavioral and neurochemical deficits in the rat 6-hydroxydopamine (6-OHDA) model of Parkinson's disease. METHODS: After unilateral injection of 6-OHDA into the median forebrain bundle, rats were assessed for behavioral function in the open field, cylinder test, and amphetamine-induced circling test. Thereafter, their brains were subjected to neurochemical and immunohistochemical analysis of dopaminergic neuron survival. Analysis was conducted as a function of treatment with P7C3 compounds, with administration initiated either before or after 6-OHDA exposure. RESULTS: Animals administered P7C3-A20 or P7C3-S243, two of the most advanced agents in the P7C3 series of neuroprotective compounds, both before and after 6-OHDA exposure showed evidence of protective efficacy in all measures. When P7C3-S243 administration was initiated after 6-OHDA exposure, rats also showed protective efficacy in all measures, which included blocking dopaminergic neuron cell death in ipsilateral substantia nigra pars compacta, preservation of dopamine and its metabolites in ipsilateral striatum, and preservation of normal motor behavior. CONCLUSIONS: The P7C3 series of compounds may form the basis for developing new therapeutic agents for slowing or preventing progression of Parkinson's disease.

17.
J Med Chem ; 57(9): 3746-54, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24697290

ABSTRACT

(-)-P7C3-S243 is a neuroprotective aminopropyl carbazole with improved druglike properties compared with previously reported compounds in the P7C3 class. It protects developing neurons in a mouse model of hippocampal neurogenesis and protects mature neurons within the substantia nigra in a mouse model of Parkinson's disease. A short, enantioselective synthesis provides the neuroprotective agent in optically pure form. It is nontoxic, orally bioavailable, metabolically stable, and able to cross the blood-brain barrier. As such, it represents a valuable lead compound for the development of drugs to treat neurodegenerative diseases and traumatic brain injury.


Subject(s)
Carbazoles/pharmacology , Neuroprotective Agents/pharmacology , Animals , Area Under Curve , Disease Models, Animal , Drug Discovery , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , Parkinson Disease/pathology , Spectrometry, Mass, Electrospray Ionization , Substantia Nigra/pathology
18.
Invest Ophthalmol Vis Sci ; 55(12): 8330-41, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25468886

ABSTRACT

PURPOSE: Traumatic brain injury (TBI) frequently leads to chronic visual dysfunction. The purpose of this study was to investigate the effect of TBI on retinal ganglion cells (RGCs), and to test whether treatment with the novel neuroprotective compound P7C3-S243 could prevent in vivo functional deficits in the visual system. METHODS: Blast-mediated TBI was modeled using an enclosed over-pressure blast chamber. The RGC physiology was evaluated using a multielectrode array and pattern electroretinogram (PERG). Histological analysis of RGC dendritic field and cell number were evaluated at the end of the study. Visual outcome measures also were evaluated based on treatment of mice with P7C3-S243 or vehicle control. RESULTS: We show that deficits in neutral position PERG after blast-mediated TBI occur in a temporally bimodal fashion, with temporary recovery 4 weeks after injury followed by chronically persistent dysfunction 12 weeks later. This later time point is associated with development of dendritic abnormalities and irreversible death of RGCs. We also demonstrate that ongoing pathologic processes during the temporary recovery latent period (including abnormalities of RGC physiology) lead to future dysfunction of the visual system. We report that modification of PERG to provocative postural tilt testing elicits changes in PERG measurements that correlate with a key in vitro measures of damage: the spontaneous and light-evoked activity of RGCs. Treatment with P7C3-S243 immediately after injury and throughout the temporary recovery latent period protects mice from developing chronic visual system dysfunction. CONCLUSIONS: Provocative PERG testing serves as a noninvasive test in the living organism to identify early damage to the visual system, which may reflect corresponding damage in the brain that is not otherwise detectable by noninvasive means. This provides the basis for developing an earlier diagnostic test to identify patients at risk for developing chronic CNS and visual system damage after TBI at an earlier stage when treatments may be more effective in preventing these sequelae. In addition, treatment with the neuroprotective agent P7C3-S243 after TBI protects from visual system dysfunction after TBI.


Subject(s)
Blast Injuries/drug therapy , Brain Injuries/drug therapy , Carbazoles/pharmacology , Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/drug effects , Vision Disorders/prevention & control , Analysis of Variance , Animals , Blast Injuries/complications , Blast Injuries/physiopathology , Brain Injuries/complications , Brain Injuries/physiopathology , Cell Count , Dendrites/pathology , Disease Models, Animal , Electroretinography/drug effects , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Vision Disorders/etiology , Vision Disorders/physiopathology
19.
Sci Transl Med ; 4(137): 137ra75, 2012 Jun 06.
Article in English | MEDLINE | ID: mdl-22674553

ABSTRACT

Most anticancer drugs entering clinical trials fail to achieve approval from the U.S. Food and Drug Administration. Drug development is hampered by the lack of preclinical models with therapeutic predictive value. Herein, we report the development and validation of a tumorgraft model of renal cell carcinoma (RCC) and its application to the evaluation of an experimental drug. Tumor samples from 94 patients were implanted in the kidneys of mice without additives or disaggregation. Tumors from 35 of these patients formed tumorgrafts, and 16 stable lines were established. Samples from metastatic sites engrafted at higher frequency than those from primary tumors, and stable engraftment of primary tumors in mice correlated with decreased patient survival. Tumorgrafts retained the histology, gene expression, DNA copy number alterations, and more than 90% of the protein-coding gene mutations of the corresponding tumors. As determined by the induction of hypercalcemia in tumorgraft-bearing mice, tumorgrafts retained the ability to induce paraneoplastic syndromes. In studies simulating drug exposures in patients, RCC tumorgraft growth was inhibited by sunitinib and sirolimus (the active metabolite of temsirolimus in humans), but not by erlotinib, which was used as a control. Dovitinib, a drug in clinical development, showed greater activity than sunitinib and sirolimus. The routine incorporation of models recapitulating the molecular genetics and drug sensitivities of human tumors into preclinical programs has the potential to improve oncology drug development.


Subject(s)
Benzimidazoles/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Quinolones/therapeutic use , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Middle Aged , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL